Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Stem Cell Res ; 64: 102919, 2022 10.
Article in English | MEDLINE | ID: mdl-36130446

ABSTRACT

The GBA gene encodes the lysosomal enzyme glucocerebrosidase (GCase), responsible for the hydrolysis of glucocerebroside to glucose and ceramide. Heterozygous GBA mutations have been associated with the development of Parkinson's disease (PD) and dementia with Lewy bodies (DLB). We generated two induced pluripotent stem cell (iPSC) lines from PD patients carrying heterozygous GBA W378G or N370S mutations and subsequently produced isogenic control lines using CRISPR/Cas9 genome editing. The patient-derived iPSCs and isogenic control lines maintained full pluripotency, normal karyotypes, and differentiation capacity. All iPSC lines could be differentiated into dopaminergic neurons, thus providing valuable tools for studying PD pathogenesis.


Subject(s)
Induced Pluripotent Stem Cells , Parkinson Disease , Humans , Glucose , Glucosylceramidase/genetics , Glucosylceramidase/metabolism , Glucosylceramides/metabolism , Induced Pluripotent Stem Cells/metabolism , Mutation/genetics , Parkinson Disease/pathology
2.
Stem Cell Res ; 62: 102806, 2022 07.
Article in English | MEDLINE | ID: mdl-35561458

ABSTRACT

Autosomal recessive mutations in either PRKN or PINK1 are associated with early-onset Parkinson's disease. The corresponding proteins, PRKN, an E3 ubiquitin ligase, and the mitochondrial serine/threonine-protein kinase PINK1 play a role in mitochondrial quality control. Using CRISPR/CAS9 technology we generated three human iPSC lines from the well characterized AIW002-02 control line. These isogenic iPSCs contain homozygous knockouts of PRKN (PRKN-KO, CBIGi001-A-1), PINK1 (PINK1-KO, CBIGi001-A-2) or both PINK1 and PRKN (PINK1-KO/PRKN-KO, CBIGi001-A-3). The knockout lines display normal karyotypes, express pluripotency markers and upon differentiation into relevant brain cells or midbrain organoids may be valuable tools to model Parkinson's disease.


Subject(s)
Induced Pluripotent Stem Cells , Parkinson Disease , CRISPR-Cas Systems/genetics , Cell Line , Humans , Induced Pluripotent Stem Cells/metabolism , Mitophagy/genetics , Parkinson Disease/genetics , Protein Kinases/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
3.
Neuroinformatics ; 20(1): 139-153, 2022 01.
Article in English | MEDLINE | ID: mdl-34003431

ABSTRACT

In January 2016, the Montreal Neurological Institute-Hospital (The Neuro) declared itself an Open Science organization. This vision extends beyond efforts by individual scientists seeking to release individual datasets, software tools, or building platforms that provide for the free dissemination of such information. It involves multiple stakeholders and an infrastructure that considers governance, ethics, computational resourcing, physical design, workflows, training, education, and intra-institutional reporting structures. The C-BIG repository was built in response as The Neuro's institutional biospecimen and clinical data repository, and collects biospecimens as well as clinical, imaging, and genetic data from patients with neurological disease and healthy controls. It is aimed at helping scientific investigators, in both academia and industry, advance our understanding of neurological diseases and accelerate the development of treatments. As many neurological diseases are quite rare, they present several challenges to researchers due to their small patient populations. Overcoming these challenges required the aggregation of datasets from various projects and locations. The C-BIG repository achieves this goal and stands as a scalable working model for institutions to collect, track, curate, archive, and disseminate multimodal data from patients. In November 2020, a Registered Access layer was made available to the wider research community at https://cbigr-open.loris.ca , and in May 2021 fully open data will be released to complement the Registered Access data. This article outlines many of the aspects of The Neuro's transition to Open Science by describing the data to be released, C-BIG's full capabilities, and the design aspects that were implemented for effective data sharing.


Subject(s)
Information Dissemination , Software , Humans
4.
Front Immunol ; 9: 834, 2018.
Article in English | MEDLINE | ID: mdl-29867923

ABSTRACT

In autoimmunity, the balance of different helper T (Th) cell subsets can influence the tissue damage caused by autoreactive T cells. Pro-inflammatory Th1 and Th17 T cells are implicated as mediators of several human autoimmune conditions such as multiple sclerosis (MS). Autologous hematopoietic stem cell transplantation (aHSCT) has been tested in phase 2 clinical trials for MS patients with aggressive disease. Abrogation of new clinical relapses and brain lesions can be seen after ablative aHSCT, accompanied by significant reductions in Th17, but not Th1, cell populations and activity. The cause of this selective decrease in Th17 cell responses following ablative aHSCT is not completely understood. We identified an increase in the kinetics of natural killer (NK) cell reconstitution, relative to CD4+ T cells, in MS patients post-aHSCT, resulting in an increased NK cell:CD4+ T cell ratio that correlated with the degree of decrease in Th17 responses. Ex vivo removal of NK cells from post-aHSCT peripheral blood mononuclear cells resulted in higher Th17 cell responses, indicating that NK cells can regulate Th17 activity. NK cells were also found to be cytotoxic to memory Th17 cells, and this toxicity is mediated through NKG2D-dependent necrosis. Surprisingly, NK cells induced memory T cells to secrete more IL-17A. This was preceded by an early rise in T cell expression of RORC and IL17A mRNA, and could be blocked with neutralizing antibodies against CD58, a costimulatory receptor expressed on NK cells. Thus, NK cells provide initial co-stimulation that supports the induction of a Th17 response, followed by NKG2D-dependent cytotoxicity that limits these cells. Together these data suggest that rapid reconstitution of NK cells following aHSCT contribute to the suppression of the re-emergence of Th17 cells. This highlights the importance of NK cells in shaping the reconstituting immune system following aHSCT in MS patients.


Subject(s)
Hematopoietic Stem Cell Transplantation , Killer Cells, Natural/immunology , Multiple Sclerosis, Relapsing-Remitting/therapy , Th17 Cells/immunology , Autoimmunity , CD58 Antigens/immunology , Cytokines/immunology , GPI-Linked Proteins/immunology , Gene Expression Regulation , Humans , Immunologic Memory , Intercellular Signaling Peptides and Proteins/immunology , Interleukin-17/immunology , Multiple Sclerosis, Relapsing-Remitting/immunology , Transplantation, Autologous
5.
Stem Cells Transl Med ; 5(11): 1506-1514, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27400792

ABSTRACT

: Human mesenchymal stem cells (hMSCs) are being increasingly pursued as potential therapies for immune-mediated conditions, including multiple sclerosis. Although they can suppress human Th1 responses, they reportedly can reciprocally enhance human Th17 responses. Here, we investigated the mechanisms underlying the capacity of hMSCs to modulate human Th1 and Th17 responses. Human adult bone marrow-derived MSCs were isolated, and their purity and differentiation capacity were confirmed. Human venous peripheral blood mononuclear cells (PBMC) were activated, alone, together with hMSC, or in the presence of hMSC-derived supernatants (sups). Cytokine expression by CD4+ T-cell subsets (intracellular staining by fluorescence-activated cell sorting) and secreted cytokines (enzyme-linked immunosorbent assay) were then quantified. The contribution of prostaglandin E2 (PGE2) as well as of myeloid cells to the hMSC-mediated regulation of T-cell responses was investigated by selective depletion of PGE2 from the hMSC sups (anti-PGE2 beads) and by the selective removal of CD14+ cells from the PBMC (magnetic-activated cell sorting separation). Human MSC-secreted products could reciprocally induce interleukin-17 expression while decreasing interferon-γ expression by human CD4+ T cells, both in coculture and through soluble products. Pre-exposure of hMSCs to IL-1ß accentuated their capacity to reciprocally regulate Th1 and Th17 responses. Human MSCs secreted high levels of PGE2, which correlated with their capacity to regulate the T-cell responses. Selective removal of PGE2 from the hMSC supernatants abrogated the impact of hMSC on the T cells. Selective removal of CD14+ cells from the PBMCs also limited the capacity of hMSC-secreted PGE2 to affect T-cell responses. Our discovery of a novel PGE2-dependent and myeloid cell-mediated mechanism by which human MSCs can reciprocally induce human Th17 while suppressing Th1 responses has implications for the use of, as well as monitoring of, MSCs as a potential therapeutic for patients with multiple sclerosis and other immune-mediated diseases. SIGNIFICANCE: Although animal studies have generated a growing interest in the anti-inflammatory potential of mesenchymal stem cells (MSCs) for the treatment of autoimmune diseases, MSCs possess the capacity to both limit and promote immune responses. Yet relatively little is known about human-MSC modulation of human disease-implicated T-cell responses, or the mechanisms underlying such modulation. The current study reveals a novel prostaglandin E2-dependent and myeloid cell-mediated mechanism by which human MSCs can reciprocally regulate human Th17 and Th1 responses, with implications for the use of MSCs as a potential therapeutic for patients with multiple sclerosis and other immune-mediated diseases.

6.
Mol Cancer ; 10: 121, 2011 Sep 24.
Article in English | MEDLINE | ID: mdl-21943176

ABSTRACT

BACKGROUND: The CCL2 chemokine is involved in promoting cancer angiogenesis, proliferation and metastasis by malignancies that express CCR2 receptor. Thus the CCL2/CCR2 axis is an attractive molecular target for anticancer drug development. METHODS: We have generated a novel fusion protein using GMCSF and an N-terminal truncated version of MCP1/CCL2 (6-76) [hereafter GMME1] and investigated its utility as a CCR2-specific tumoricidal agent. RESULTS: We found that distinct to full length CCL2 or its N-truncated derivative (CCL2 5-76), GMME1 bound to CCR2 on mouse lymphoma EG7, human multiple myeloma cell line U266, or murine and human medulloblastoma cell lines, and led to their death by apoptosis. We demonstrated that GMME1 specifically blocked CCR2-associated STAT3 phosphorylation and up-regulated pro-apoptotic BAX. Furthermore, GMME1 significantly inhibited EG7 tumor growth in C57BL/6 mice, and induced apoptosis of primary myeloma cells from patients. CONCLUSION: Our data demonstrate that GMME1 is a fusokine with a potent, CCR2 receptor-mediated pro-apoptotic effect on tumor cells and could be exploited as a novel biological therapy for CCR2+ malignancies including lymphoid and central nervous system malignancies.


Subject(s)
Antineoplastic Agents/pharmacology , Chemokine CCL2/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, CCR2/metabolism , Recombinant Fusion Proteins/pharmacology , Animals , Antigens, CD/metabolism , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Female , Humans , Lymphoma , Medulloblastoma , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/therapeutic use , Tumor Burden , Xenograft Model Antitumor Assays
7.
Expert Rev Neurother ; 11(9): 1295-303, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21864075

ABSTRACT

Phase I clinical trials exploring the use of autologous mesenchymal stem cell (MSC) therapy for the treatment of multiple sclerosis (MS) have begun in a number of centers across the world. MS is a complex and chronic immune-mediated and neurodegenerative disease influenced by genetic susceptibility and environmental risk factors. The ideal treatment for MS would involve both attenuation of detrimental inflammatory responses, and induction of a degree of tissue protection/regeneration within the CNS. Preclinical studies have demonstrated that both human-derived and murine-derived MSCs are able to improve outcomes in the animal model of MS, experimental autoimmune encephalomyelitis. How MSCs ameliorate experimental autoimmune encephalomyelitis is being intensely investigated. One of the major mechanisms of action of MSC therapy is to inhibit various components of the immune system that contribute to tissue destruction. Emerging evidence now supports the idea that MSCs can access the CNS where they can provide protection against tissue damage, and may facilitate tissue regeneration through the production of growth factors. The prospect of cell-based therapy using MSCs has several advantages, including the relative ease with which they can be extracted from autologous bone marrow or adipose tissue and expanded in vitro to reach the purity and numbers required for transplantation, and the fact that MSC therapy has already been used in other human disease settings, such as graft-versus-host and cardiac disease, with initial reports indicating a good safety profile. This article will focus on the theoretical and practical issues relevant to considerations of MSC therapy in the context of MS.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology , Multiple Sclerosis/therapy , Animals , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Encephalomyelitis, Autoimmune, Experimental/therapy , Humans , Mice , Multiple Sclerosis/immunology , Multiple Sclerosis/physiopathology
8.
J Immunol ; 185(12): 7358-66, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21076067

ABSTRACT

We have previously shown that the fusion of GM-CSF and IL-21 (GIFT-21) possesses a potent immune stimulatory effect on myeloid cells. In this study, we define the effect of GIFT-21 on naive murine monocytes (GIFT-21 dendritic cells [DCs]), which express increased levels of Gr-1, CD45R, MHC class I, CD80, CD86, and CXCR4 and suppress CD11c and MHC class II. Compared with conventional dendritic cells, GIFT-21 DCs produced substantially more CCL2, IL-6, TNF-α, and IFN-α and induced significantly greater production of IFN-γ by CD8(+) T cells in MHC class I-restricted Ag presentation assays. B16 melanoma and D2F2 Neu breast cancer growth was inhibited in mice treated with Ag-naive GIFT-21 DCs. This effect was lost in CD8(-/-) and CCR2(-/-) mice and when mice were treated with ß(2)-microglobulin-deficient GIFT-21 DCs, indicating that GIFT-21 DCs migrated to and sampled from the tumors to present tumor Ags to CCL2 recruited CD8(+) T cells via MHC class I. We propose that autologous GIFT-21 DCs may serve as a cell therapy platform for the treatment of cancer.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Immunity, Cellular/drug effects , Interleukins/immunology , Mammary Neoplasms, Experimental/immunology , Melanoma/immunology , Recombinant Fusion Proteins/pharmacology , Adoptive Transfer , Animals , Antigen Presentation/drug effects , Antigen Presentation/genetics , Antigen Presentation/immunology , Antigens, Differentiation/genetics , Antigens, Differentiation/immunology , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Movement/immunology , Cytokines/genetics , Cytokines/immunology , Dendritic Cells/transplantation , Female , Immunity, Cellular/genetics , Immunity, Cellular/immunology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/therapy , Melanoma/genetics , Melanoma/therapy , Mice , Mice, Inbred BALB C , Mice, Knockout , Transplantation, Autologous
9.
Cancer Res ; 70(20): 7742-7, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20924101

ABSTRACT

It is unknown whether mesenchymal stromal cells (MSC) can regulate immune responses targeting tumor autoantigens of low immunogenicity. We tested here whether immunization with MSC could break immune tolerance towards the ErbB-2/HER-2/neu tumor antigen and the effects of priming with IFN-γ and tumor necrosis factor-α (TNF-α) on this process. BALB/c- and C57BL/6-derived MSC were lentivirally transduced to express a kinase-inactive rat neu mutant (MSC/Neu). Immunization of BALB/c mice with nontreated or IFN-γ-primed allogeneic or syngeneic MSC/Neu induced similar levels of anti-neu antibody titers; however, only syngeneic MSC/Neu induced protective neu-specific CD8(+) T cell responses. Compared to immunization with nontreated or IFN-γ-primed syngeneic MSC/Neu, the number of circulating neu-specific CD8(+) T cells and titers of anti-neu antibodies were observed to be decreased after immunizations with IFN-γ- plus TNF-α-primed MSC/Neu. In addition, syngeneic MSC/Neu seemed more efficient than IFN-γ-primed MSC/Neu at inducing a protective therapeutic antitumor immune response resulting in the regression of transplanted neu-expressing mammary tumor cells. In vitro antigen-presenting cell assays performed with paraformaldehyde-fixed or live MSC showed that priming with IFN-γ plus TNF-α, compared to priming with IFN-γ alone, increased antigen presentation as well as the production of immunosuppressive factors. These data suggest that whereas MSC could effectively serve as antigen-presenting cells to induce immune responses aimed at tumor autoantigens, these functions are critically regulated by IFN-γ and TNF-α.


Subject(s)
Breast Neoplasms/immunology , Interferon-gamma/therapeutic use , Mammary Neoplasms, Experimental/immunology , Mesenchymal Stem Cells/immunology , Receptor, ErbB-2/biosynthesis , Tumor Necrosis Factor-alpha/therapeutic use , Animals , Breast Neoplasms/therapy , Cancer Vaccines/therapeutic use , Female , Humans , Mammary Neoplasms, Experimental/pathology , Mammary Tumor Virus, Mouse/genetics , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Promoter Regions, Genetic , Rats , Stromal Cells/immunology , Stromal Cells/pathology
10.
Am J Physiol Renal Physiol ; 299(6): F1288-98, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20844023

ABSTRACT

Acute kidney injury (AKI) can occur from the toxic side-effects of chemotherapeutic agents such as cisplatin. Bone marrow-derived mesenchymal stromal cells (MSCs) have demonstrated wide therapeutic potential often due to beneficial factors they secrete. The goal of this investigation was to evaluate in vitro the effect of human MSCs (hMSCs) secretome on cisplatin-treated human kidney cells, and in vivo the consequence of hMSCs intraperitoneal (ip) implantation in mice with AKI. Our results revealed that hMSCs-conditioned media improved survival of HK-2 human proximal tubular cells exposed to cisplatin in vitro. This enhanced survival was linked to increased expression of phosphorylated Akt (Ser473) and was reduced by a VEGF-neutralizing antibody. In vivo testing of these hMSCs established that ip administration in NOD-SCID mice decreased cisplatin-induced kidney function impairment, as demonstrated by lower blood urea nitrogen levels and higher survival. In addition, blood phosphorous and amylase levels were also significantly decreased. Moreover, hMSCs reduced the plasma levels of several inflammatory cytokines/chemokines. Immunohistochemical examination of kidneys showed less apoptotic and more proliferating cells. Furthermore, PCR indicated the presence of hMSCs in mouse kidneys, which also showed enhanced expression of phosphorylated Akt. In conclusion, our study reveals that hMSCs can exert prosurvival effects on renal cells in vitro and in vivo, suggests a paracrine contribution for kidney protective abilities of hMSCs delivered ip, and supports their clinical potential in AKI.


Subject(s)
Acute Kidney Injury/therapy , Cisplatin/adverse effects , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology , Acute Kidney Injury/chemically induced , Acute Kidney Injury/pathology , Animals , Cell Proliferation/drug effects , Cells, Cultured , Culture Media, Conditioned/pharmacology , Cytokines/blood , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Proto-Oncogene Proteins c-akt/metabolism , Stromal Cells/physiology
11.
Ann Neurol ; 68(4): 540-5, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20661924

ABSTRACT

Human mesenchymal stem cells (hMSCs) are being considered for clinical trials of multiple sclerosis (MS). We examined the effects of adult bone marrow-derived hMSCs on responses of primary human Th1, Th17, and Th1/17 double-expressing T-cell subsets, all implicated in MS. As expected, soluble products from hMSCs inhibited Th1 responses; however, Th17 responses were increased. Secretion of interleukin (IL)-10, considered anti-inflammatory, was decreased. Pretreating hMSCs with the proinflammatory cytokine IL-1ß accentuated these effects, and caused decreases in the Th1/17 subset. These findings underscore the importance of further preclinical work and immune-monitoring to define hMSC effects on disease-relevant immune responses under variable conditions.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Mesenchymal Stem Cells/immunology , Th1 Cells/cytology , Antigens, CD/metabolism , CD4-Positive T-Lymphocytes/classification , CD4-Positive T-Lymphocytes/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Culture Media, Conditioned/pharmacology , Cytokines/immunology , Cytokines/pharmacology , Enzyme-Linked Immunosorbent Assay/methods , Flow Cytometry/methods , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Lymphocyte Activation/immunology , Mesenchymal Stem Cells/chemistry , T-Lymphocytes, Helper-Inducer/classification , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , Th1 Cells/immunology
12.
Nat Med ; 15(9): 1038-45, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19668193

ABSTRACT

We have previously shown that a granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-15 (IL-15) 'fusokine' (GIFT15) exerts immune suppression via aberrant signaling through the IL-15 receptor on lymphomyeloid cells. We show here that ex vivo GIFT15 treatment of mouse splenocytes generates suppressive regulatory cells of B cell ontogeny (hereafter called GIFT15 B(reg) cells). Arising from CD19+ B cells, GIFT15 B(reg) cells express major histocompatibility complex class I (MHCI) and MHCII, surface IgM and IgD, and secrete IL-10, akin to previously described B10 and T2-MZP B(reg) cells, but lose expression of the transcription factor PAX5, coupled to upregulation of CD138 and reciprocal suppression of CD19. Mice with experimental autoimmune encephalomyelitis went into complete remission after intravenous infusion of GIFT15 B(reg) cells paralleled by suppressed neuroinflammation. The clinical effect was abolished when GIFT15 B(reg) cells were derived from mmicroMT (lacking B cells), MHCII-knockout, signal transducer and activator of transcription-6 (STAT-6)-knockout, IL-10-knockout or allogeneic splenocytes, consistent with a pivotal role for MHCII and IL-10 by sygeneic B cells for the observed therapeutic effect. We propose that autologous GIFT15 B(reg) cells may serve as a new treatment for autoimmune ailments.


Subject(s)
B-Lymphocyte Subsets/drug effects , B-Lymphocyte Subsets/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Interleukin-15/pharmacology , Recombinant Fusion Proteins/pharmacology , Animals , B-Lymphocyte Subsets/transplantation , Cytokines , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/therapy , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class II/metabolism , Immune Tolerance/drug effects , In Vitro Techniques , Interferon-gamma/biosynthesis , Interleukin-10/biosynthesis , Mice , Mice, Knockout , Mice, Transgenic , Recombinant Proteins , STAT6 Transcription Factor/deficiency , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/immunology , Transplantation, Homologous , Transplantation, Isogeneic
13.
Blood ; 114(13): 2632-8, 2009 Sep 24.
Article in English | MEDLINE | ID: mdl-19654411

ABSTRACT

Recent studies involving bone marrow mesenchymal stromal cells (MSCs) demonstrated that interferon (IFN)-gamma stimulation induces major histocompatibility complex (MHC) class II-mediated antigen presentation in MSCs both in vitro and in vivo. Concordantly, we investigated the ability of MSCs to present extracellular antigen through their MHC class I molecules, a process known as cross-presentation. Using an in vitro antigen presentation assay, we demonstrated that murine MSCs can cross-present soluble ovalbumin (OVA) to naive CD8(+) T cells from OT-I mice. Cross-presentation by MSC was proteasome dependent and partly dependent on transporter associated with antigen-processing molecules. Pretreatment of MSC with IFN-gamma increased cross-presentation by up-regulating antigen processing and presentation. However, although the transcription of the transporter associated with antigen processing-1 molecules and the immunoproteasome subunit LMP2 induced by IFN-gamma was inhibited by transforming growth factor-beta, the overall cross-presentation capacity of MSCs remained unchanged after transforming growth factor-beta treatment. These observations were validated in vivo by performing an immune reconstitution assay in beta(2)-microglobulin(-/-) mice and show that OVA cross-presentation by MSCs induces the proliferation of naive OVA-specific CD8(+) T cells. In conclusion, we demonstrate that MSCs can cross-present exogenous antigen and induce an effective CD8(+) T-cell immune response, a property that could be exploited as a therapeutic cell-based immune biopharmaceutic for the treatment of cancer or infectious diseases.


Subject(s)
Antigen-Presenting Cells/immunology , Antigens/immunology , Cross-Priming/immunology , Mesenchymal Stem Cells/immunology , Stromal Cells/immunology , Animals , Antigen Presentation/immunology , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/physiology , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Female , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/immunology , Solubility , Stromal Cells/metabolism , Stromal Cells/physiology , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/physiology , beta 2-Microglobulin/genetics
14.
Blood ; 114(15): 3181-90, 2009 Oct 08.
Article in English | MEDLINE | ID: mdl-19587377

ABSTRACT

Gaucher disease causes pathologic skeletal changes that are not fully explained. Considering the important role of mesenchymal stromal cells (MSCs) in bone structural development and maintenance, we analyzed the cellular biochemistry of MSCs from an adult patient with Gaucher disease type 1 (N370S/L444P mutations). Gaucher MSCs possessed a low glucocerebrosidase activity and consequently had a 3-fold increase in cellular glucosylceramide. Gaucher MSCs have a typical MSC marker phenotype, normal osteocytic and adipocytic differentiation, growth, exogenous lactosylceramide trafficking, cholesterol content, lysosomal morphology, and total lysosomal content, and a marked increase in COX-2, prostaglandin E2, interleukin-8, and CCL2 production compared with normal controls. Transcriptome analysis on normal MSCs treated with the glucocerebrosidase inhibitor conduritol B epoxide showed an up-regulation of an array of inflammatory mediators, including CCL2, and other differentially regulated pathways. These cells also showed a decrease in sphingosine-1-phosphate. In conclusion, Gaucher disease MSCs display an altered secretome that could contribute to skeletal disease and immune disease manifestations in a manner distinct and additive to Gaucher macrophages themselves.


Subject(s)
Bone Marrow Cells/metabolism , Chemokine CCL2/metabolism , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Gaucher Disease/metabolism , Inflammation Mediators/metabolism , Interleukin-8/metabolism , Adipocytes/metabolism , Adipocytes/pathology , Amino Acid Substitution , Bone Marrow Cells/pathology , Cell Differentiation/drug effects , Cells, Cultured , Chemokine CCL2/biosynthesis , Chemokine CCL2/genetics , Cholesterol/genetics , Cholesterol/metabolism , Cyclooxygenase 2/biosynthesis , Cyclooxygenase 2/genetics , Dinoprostone/biosynthesis , Dinoprostone/genetics , Enzyme Inhibitors/pharmacology , Female , Gaucher Disease/genetics , Gaucher Disease/pathology , Glucosylceramidase/antagonists & inhibitors , Glucosylceramidase/genetics , Glucosylceramidase/metabolism , Glucosylceramides/genetics , Glucosylceramides/metabolism , Humans , Inositol/analogs & derivatives , Inositol/pharmacology , Interleukin-8/biosynthesis , Interleukin-8/genetics , Lysosomes/metabolism , Lysosomes/pathology , Male , Middle Aged , Mutation, Missense , Osteocytes/metabolism , Osteocytes/pathology , Stromal Cells/metabolism , Stromal Cells/pathology , Up-Regulation/drug effects , Up-Regulation/genetics
15.
J Immunol ; 183(3): 1759-66, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19592643

ABSTRACT

CCR2 is a chemokine receptor widely expressed by lymphomyeloid cells involved in maladaptive autoimmune ailments. Therefore CCR2 is of great interest as a biological target for immune suppression due to its direct implication in autoimmune diseases such as rheumatoid arthritis. We have generated a novel fusion protein using GM-CSF and an N-terminal truncated version of MCP-1/CCL2 (6-76, GMME1) and investigated its utility as a CCR2-specific immune suppressor. Using BRET studies, we found that distinct to CCL2, GMME1 binding to CCR2 led to altered conformational changes in the CCR2 homodimer and did not induce the recruitment of beta-arrestin 2 to the receptor. However, CCR2-dependent calcium mobilization, BAX induction and caspase-3 activation followed by cell death was observed. Using Th17 cells harvested from DBA/1 mice ill with bovine collagen-induced arthritis, we demonstrate that GMME1 is capable of blocking their production of IL-17 in vitro. Upon its delivery to mice symptomatic with inflammatory arthritis, a robust clinical recovery occurred with decreased paw thickness to normal levels and a significant reduction in anti-collagen Ab titer and rheumatoid factor titer, as well as reduction of proinflammatory cytokines levels both intraarticular and systemic. Our data demonstrate that GMME1 is a powerful synthetic suppressor cytokine that coopts CCR2-dependent cellular signaling and blunts the effects of CCR2-expressing lymphomyeloid cells causative of autoimmune arthritis.


Subject(s)
Arthritis, Experimental/drug therapy , Chemokine CCL2/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Protein Engineering/methods , Receptors, CCR2/metabolism , Recombinant Fusion Proteins/pharmacology , Animals , Arthritis, Experimental/prevention & control , Cells, Cultured , Disease Models, Animal , Inflammation/drug therapy , Inflammation/prevention & control , Interleukin-17 , Mice , Protein Binding , Receptors, CCR2/chemistry , Recombinant Fusion Proteins/therapeutic use , T-Lymphocyte Subsets
16.
J Immunol ; 182(12): 7963-73, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19494321

ABSTRACT

Bone marrow-derived mesenchymal stromal cells (MSC) possess an immune plasticity manifested by either an immunosuppressive or, when activated with IFN-gamma, an APC phenotype. Herein, TLR expression by MSC and their immune regulatory role were investigated. We observed that human MSC and macrophages expressed TLR3 and TLR4 at comparable levels and TLR-mediated activation of MSC resulted in the production of inflammatory mediators such as IL-1beta, IL-6, IL-8/CXCL8, and CCL5. IFN-alpha or IFN-gamma priming up-regulated production of these inflammatory mediators and expression of IFNB, inducible NO synthase (iNOS), and TRAIL upon TLR activation in MSC and macrophages, but failed to induce IL-12 and TNF-alpha production in MSC. Nonetheless, TLR activation in MSC resulted in the formation of an inflammatory site attracting innate immune cells, as evaluated by human neutrophil chemotaxis assays and by the analysis of immune effectors retrieved from Matrigel-embedded MSC injected into mice after in vitro preactivation with cytokines and/or TLR ligands. Hence, TLR-activated MSC are capable of recruiting immune inflammatory cells. In addition, IFN priming combined with TLR activation may increase immune responses induced by Ag-presenting MSC through presentation of Ag in an inflammatory context, a mechanism that could be applied in a cell-based vaccine.


Subject(s)
Cytokines/immunology , Cytokines/metabolism , Stromal Cells/immunology , Stromal Cells/metabolism , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 4/metabolism , Adult , Animals , Cells, Cultured , Female , Gene Expression Regulation , Humans , Inflammation/immunology , Inflammation/metabolism , Interferon Regulatory Factor-1/metabolism , Ligands , Ligases/metabolism , Male , Mice , Phenotype , Proto-Oncogene Proteins c-rel/metabolism , Toll-Like Receptor 3/genetics , Toll-Like Receptor 4/genetics
17.
J Immunol ; 182(10): 5994-6002, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19414750

ABSTRACT

The administration of ex vivo culture-expanded mesenchymal stromal cells (MSCs) has been shown to reverse symptomatic neuroinflammation observed in experimental autoimmune encephalomyelitis (EAE). The mechanism by which this therapeutic effect occurs remains unknown. In an effort to decipher MSC mode of action, we found that MSC conditioned medium inhibits EAE-derived CD4 T cell activation by suppressing STAT3 phosphorylation via MSC-derived CCL2. Further analysis demonstrates that the effect is dependent on MSC-driven matrix metalloproteinase proteolytic processing of CCL2 to an antagonistic derivative. We also show that antagonistic CCL2 suppresses phosphorylation of AKT and leads to a reciprocal increased phosphorylation of ERK associated with an up-regulation of B7.H1 in CD4 T cells derived from EAE mice. CD4 T cell infiltration of the spinal cord of MSC-treated group was robustly decreased along with reduced plasma levels of IL-17 and TNF-alpha levels and in vitro from restimulated splenocytes. The key role of MSC-derived CCL2 was confirmed by the observed loss of function of CCL2(-/-) MSCs in EAE mice. In summary, this is the first report of MSCs modulating EAE biology via the paracrine conversion of CCL2 from agonist to antagonist of CD4 Th17 cell function.


Subject(s)
Chemokine CCL2/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Mesenchymal Stem Cells/immunology , Stromal Cells/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Blotting, Western , Chemokine CCL2/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Flow Cytometry , Interleukin-17/immunology , Interleukin-17/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Stromal Cells/metabolism , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Helper-Inducer/metabolism
18.
J Immunol ; 182(5): 2620-7, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19234156

ABSTRACT

We describe the generation of a fusion cytokine consisting of GM-CSF in tandem with N-terminal-truncated MCP-1 (6-76), hereafter GMME1. Treatment of activated T cells with recombinant GMME1 protein leads to proinflammatory cytokine reduction and apoptosis via a CCR2-restricted pathway. Similarly, cell death is triggered in macrophages cultured with GMME1, while an inhibition of Ab production from plasma cells is observed. Treatment of CD4 T cells derived from experimental autoimmune encephalomyelitis mice with GMME1 leads to p38 hyperphosphorylation, inhibition of p44/42, AKT and STAT3 phosphorylation, and caspase-3 activation. GMME1 administration to experimental autoimmune encephalomyelitis mice suppresses symptomatic disease and correlates with decreased levels of inflammatory cytokines including IL-17, MOG-specific Ab titers, and blockade of CD4 and CD8 T cell infiltration in spinal cords. We propose that GMME1 defines a new class of agents for the treatment of autoimmune ailments by selectively targeting lymphomyeloid cells expressing CCR2.


Subject(s)
Chemokine CCL2/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Lymphocytes/immunology , Myeloid Cells/immunology , Receptors, CCR2/antagonists & inhibitors , Receptors, CCR2/genetics , Recombinant Fusion Proteins/physiology , Amino Acid Sequence , Animals , Apoptosis/genetics , Apoptosis/immunology , Cells, Cultured , Chemokine CCL2/administration & dosage , Chemokine CCL2/therapeutic use , Coculture Techniques , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/therapy , Female , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , HeLa Cells , Humans , Lymphocytes/metabolism , Lymphocytes/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Myeloid Cells/metabolism , Myeloid Cells/pathology , Receptors, CCR2/biosynthesis , Receptors, CCR2/deficiency , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/chemical synthesis
19.
Blood ; 112(13): 4991-8, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18812467

ABSTRACT

We demonstrate that the secretome of mesenchymal stromal cells (MSCs) suppresses plasma cell (PC) immunoglobulin (Ig) production, induces plasmablast proliferation, and leads to interleukin-10-mediated blockade in vitro. We found that these effects are the result of MSC-derived CC chemokine ligands CCL2 and CCL7. More specifically, MSCs further processed these CC chemokines by the activity of matrix metalloproteinases (MMPs), leading to the generation of proteolytically processed antagonistic CCL2 variant. Neutralizing CCL2 or inhibiting MMP enzymatic activity abolished the PC-suppressive effect of MSCs. We also observed that MMP-processed CCL2 suppresses signal transducer and activator of transcription 3 (STAT3) activation in PC. As a result, the transcription factor PAX5 is induced, thus explaining the inhibition of Ig synthesis. The absence of inhibitory effects by MSC on the humoral response of CCR2(-/-) mice to xenoantigen suggests that MMP-cleaved CCL2/CCR2 interaction as well as downstream phosphatase activity is necessary for antagonistic effect. We tested syngeneic MSCs in hemophilic B6 mice with predeveloped antihuman factor VIII (hFVIII) antibodies and demonstrated a robust decrease in hFVIII-specific IgG levels. Thus, MSCs may play a role in modulating Ig production by PCs via MMP processing of CCL2 and may represent an appealing cell therapy approach for pathologic humoral responses.


Subject(s)
Chemokine CCL2/immunology , Immunoglobulins/biosynthesis , Mesenchymal Stem Cells/immunology , PAX5 Transcription Factor/genetics , Plasma Cells/metabolism , STAT3 Transcription Factor/metabolism , Stromal Cells/immunology , Animals , Chemokine CCL7/immunology , Matrix Metalloproteinases/metabolism , Mice , Plasma Cells/immunology , Transcriptional Activation/immunology
20.
Cancer Res ; 68(12): 4810-8, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18559528

ABSTRACT

Bone marrow-derived mesenchymal stromal cells (MSCs), beneficial for regenerative medicine applications due to their wide differentiation capabilities, also hold promise as cellular vehicles for the delivery of therapeutic plasma-soluble gene products due to their ease of handling, expansion, and genetic engineering. We hypothesized that MSCs, gene enhanced to express interleukin-12 (IL-12) and then embedded in a matrix, may act as an anticancer neo-organoid when delivered s.c. in autologous/syngeneic hosts. We performed such experiments in mice and noted that primary murine MSCs retrovirally engineered to secrete murine IL-12 can significantly interfere with growth of 4T1 breast cancer cells in vivo, with a more substantial anticancer action achieved when these cells are embedded in a matrix. Plasma of mice that received the IL-12 MSC-containing neo-organoids showed increased levels of IL-12 and IFN-gamma. Histopathologic analysis revealed less tumor cells in implants of 4T1 cells with IL-12 MSCs, and the presence of necrotic tumor islets and necrotic capillaries, suggesting antiangiogenesis. We also showed that the anticancer effect exerted by the IL-12 MSCs is immune mediated because it is absent in immunodeficient mice, is not due to systemic IL-12 delivery, and also occurs in a B16 melanoma model. This study therefore establishes the feasibility of using gene-enhanced MSCs in a cell-based neo-organoid approach for cancer treatment.


Subject(s)
Bone Marrow Cells/metabolism , Immunotherapy , Interleukin-12/therapeutic use , Mammary Neoplasms, Experimental/therapy , Mesoderm/cytology , Organoids , Stromal Cells/immunology , Animals , Cells, Cultured , Female , Interleukin-12/metabolism , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/metabolism , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/therapy , Mesoderm/immunology , Mesoderm/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Neovascularization, Physiologic , Stromal Cells/metabolism , Stromal Cells/pathology , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL
...